Skip to main content
Taylor & Francis Open Select logoLink to Taylor & Francis Open Select
. 2015 Sep 18;36(6):1110–1122. doi: 10.3109/07388551.2015.1084266

Human cell lines for biopharmaceutical manufacturing: history, status, and future perspectives

Jennifer Dumont a,*, Don Euwart a, Baisong Mei a, Scott Estes a, Rashmi Kshirsagar a
PMCID: PMC5152558  PMID: 26383226

Abstract

Biotherapeutic proteins represent a mainstay of treatment for a multitude of conditions, for example, autoimmune disorders, hematologic disorders, hormonal dysregulation, cancers, infectious diseases and genetic disorders. The technologies behind their production have changed substantially since biotherapeutic proteins were first approved in the 1980s. Although most biotherapeutic proteins developed to date have been produced using the mammalian Chinese hamster ovary and murine myeloma (NS0, Sp2/0) cell lines, there has been a recent shift toward the use of human cell lines. One of the most important advantages of using human cell lines for protein production is the greater likelihood that the resulting recombinant protein will bear post-translational modifications (PTMs) that are consistent with those seen on endogenous human proteins. Although other mammalian cell lines can produce PTMs similar to human cells, they also produce non-human PTMs, such as galactose-α1,3-galactose and N-glycolylneuraminic acid, which are potentially immunogenic. In addition, human cell lines are grown easily in a serum-free suspension culture, reproduce rapidly and have efficient protein production. A possible disadvantage of using human cell lines is the potential for human-specific viral contamination, although this risk can be mitigated with multiple viral inactivation or clearance steps. In addition, while human cell lines are currently widely used for biopharmaceutical research, vaccine production and production of some licensed protein therapeutics, there is a relative paucity of clinical experience with human cell lines because they have only recently begun to be used for the manufacture of proteins (compared with other types of cell lines). With additional research investment, human cell lines may be further optimized for routine commercial production of a broader range of biotherapeutic proteins.

Keywords: Cell culture, clotting factor, glycosylation, HEK 293, immunogenicity, monoclonal antibody, post-translational modification, recombinant protein, therapeutic glycoprotein, vaccine

Introduction

Protein therapeutics (including monoclonal antibodies [mAbs], peptides and recombinant proteins) represent the largest group of new products in development by the biopharmaceutical industry (Durocher & Butler, 2009; Ho & Chien, 2014).

These products are produced in a wide variety of platforms, including non-mammalian expression systems (bacterial, yeast, plant and insect) and mammalian expression systems (including human cell lines) (Ghaderi et al., 2012). Importantly, the most appropriate expression system depends on the particular protein to be expressed. Mammalian expression systems are generally the preferred platform for manufacturing biopharmaceuticals, as these cell lines are able to produce large, complex proteins with post-translational modifications (PTMs; most notably glycosylation) similar to those produced in humans (Durocher & Butler, 2009; Ghaderi et al., 2012; Swiech et al., 2012). Moreover, in the case of mammalian cell lines, and animal cell lines in general, most proteins can be secreted rather than requiring cell lysis to extract with subsequent protein refolding (as is the case with bacteria/prokaryotes). The most common mammalian (non-human) cell lines used for therapeutic protein production include Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK21) cells and murine myeloma cells (NS0 and Sp2/0) (Estes & Melville, 2014). However, these non-human mammalian cell lines also produce PTMs that are not expressed in humans, namely galactose-α1,3-galactose (α-gal) and N-glycolylneuraminic acid (NGNA). Because humans possess circulating antibodies against both of these N-glycans, non-human cell lines are usually screened during their production to identify clones with acceptable glycan profiles (Ghaderi et al., 2010).

Human cell lines have the ability to produce proteins most similar to those synthesized naturally in humans, which may be an advantage compared with other mammalian expression systems (Ghaderi et al., 2010). In particular, the structure, number and location of post-translational N-glycans can affect the biologic activity, protein stability, clearance rate and immunogenicity of biotherapeutic proteins (Arnold et al., 2007; Ghaderi et al., 2010; Swiech et al., 2012).

The first human cell line, HeLa, was established in 1951 from a cervical cancer (Scherer et al., 1953). Human diploid cells were developed in the 1960s for vaccine manufacturing; however, concerns for a latent oncogenic agent in these cell lines (despite a lack of suggestive phenotypic characteristics) delayed their acceptance. Currently, human diploid cells are used in the manufacture of many viral vaccines (Petricciani & Sheets, 2008). However, due to their rapid growth, high protein yield, and the investment in system optimization, animal cells remained the substrate of choice for the production of recombinant proteins and mAbs (Petricciani & Sheets, 2008).

Today, advances in technology have allowed for increased productivity with human cell lines, and there are now approved recombinant biotherapeutic products produced from the human embryonic kidney 293 (HEK293) and fibrosarcoma HT-1080 cell lines (Beck, 2009; Casademunt et al., 2012; Dumont et al., 2012; Glaesner et al., 2010; Peters et al., 2010; Wraith, 2008; Zimran et al., 2013). Additional biotherapeutic products produced in the PER.C6, HKB-11, CAP and HuH-7 human cell lines are currently being evaluated (Enjolras et al., 2012; Estes & Melville, 2014; Jones et al., 2003; Mei et al., 2006; Swiech et al., 2011, 2015). This article is a narrative review of the cell lines (with a focus on human cell lines) used for production of biotherapeutic proteins, both approved and in development.

Non-human expression systems used to manufacture biotherapeutic products

Many non-human expression systems have been utilized in the production of currently approved biotherapeutic proteins (Table 1).

Table 1. Non-human expression systems used in the production of biotherapeutics approved in the United States and Europea,b.

Expression system Biotherapeutic product FDA approval EMA approval
Plant cells Enzymes    
   Taliglucerase alfa Approved NA
Insect cells Vaccines    
   Cervical cancer vaccine Approved Approved
Bacteria Monoclonal antibodies    
   Certolizumab pegol Approved Approved
  Cytokines    
   tbo-filgrastim Approved NA
   Romiplostim Approved Approved
  Enzymes    
   Asparaginase Erwinia chrysanthemi Approved NA
   Glucarpidase Approved NA
   Pegloticase Approved Approved
   Collagenase Clostridium histolyticum Approved NA
  Peptides Approved NA
   Metreleptin    
  Therapeutic toxins Approved NA
   Incobotulinumtoxin A    
   Vaccines    
   Meningitis vaccine Approved Approved
   Pneumococcal vaccine Approved Approved
Yeast Enzymes    
   Ocriplasmin Approved Approved
  Peptides    
   Albiglutide Approved Approved
   Liraglutide Approved Approved
  Clotting factors    
   Catridecacog Approved Approved
Mammalian (non-human) cell lines    
CHO Monoclonal antibodies    
   Adalimumab Approved Approved
   Alemtuzumab Approved NA
   Bevacizumab Approved Approved
   Brentuximab vedotin Approved Approved
   Denosumab Approved Approved
   Golimumab Approved Approved
   Ibritumomab tiuxetan Approved Approved
   Ipilimumab Approved Approved
   Obinutuzumab Approved Approved
   Omalizumab Approved Approved
   Panitumumab Approved Approved
   Pertuzumab Approved Approved
   Rituximab Approved Approved
   Siltuximab Approved Approved
   Tocilizumab Approved Approved
   Trastuzumab Approved Approved
   Vedolizumab Approved Approved
   Ado-trastuzumabemtansine Approved Approved
   Ustekinumab Approved Approved
  Cytokines    
   Darbepoetin alfa Approved Approved
   Interferon beta-1a Approved Approved
   Epoetin alfa Approved Approved
   Epoetin beta NA NA
   Epoetin theta NA Approved
  Enzymes    
   Agalsidase beta Approved Approved
   Alglucosidase alfa Approved Approved
   Alteplase Approved Approved
   Elosulfase alfa Approved NA
   GalNAc 4-sulfatase Approved NA
   Human DNase Approved Approved
   Hyaluronidase Approved NA
   Imiglucerase Approved NA
   Laronidase Approved NA
   Tenecteplase Approved Approved
  Fc-fusion proteins    
   Abatacept Approved Approved
   Aflibercept Approved Approved
   Alefacept Approved Approved
   Belatacept Approved Approved
   Etanercept Approved NA
   Rilonacept Approved Approved
   Ziv-aflibercept Approved Approved
  Hormones    
   Choriogonadotropin alfa Approved NA
   Follitropin alfa Approved Approved
   Follitropin beta Approved Approved
   Luteinizing hormone Approved Approved
   Osteogenic protein-1 Approved Approved
   Thyrotropin alfa Approved Approved
  Clotting factors    
   Factor VIII Approved Approved
   Factor IX Approved Approved
NS0 Monoclonal antibodies    
   Belimumab Approved Approved
   Natalizumab Approved Approved
   Ofatumumab Approved Approved
   Palivizumab Approved Approved
   Ramucirumab Approved NA
Sp2/0 Monoclonal antibodies    
   Abciximab Approved NA
   Basiliximab Approved Approved
   Canakinumab Approved Approved
   Cetuximab Approved Approved
   Infliximab Approved Approved
BHK Clotting factors    
   Factor VIIa Approved Approved
   Factor VIII Approved Approved
Murine C127 Hormones    
   Somatropin Approved Approved

FDA, US Food and Drug Administration; EMA, European Medicines Agency; NA, not approved; CHO, Chinese hamster ovary; BHK, baby hamster kidney.

aData obtained from publically available resources (October 2014); all approved products may not be included.

bReferences: (ABSEAMED®, 2012; ACTEMRA®, 2013; ACTILYSE®, 2014; ACTIVASE®, 2012; ADCETRIS®, 2013; ADVATE®, 2014; ALDURAZYME®, 2008; ARANESP®, 2006; ARCALYST™, 2008; ARZERRA®, 2014; AVASTIN®, 2010; AVONEX®, 2007; BENEFIX®, 2012; BENLYSTA®, 2011; CAMPATH®, 2014; CATHFLO® ACTIVASE®, 2010; CEREZYME®, 2010; CERVARIX®, 2012; CIMZIA®, 2013; CIMZIA®, 2014; CYRAMZA®, 2014; ELELYSO™, 2014; ENBREL®, 2010; ENTYVIO®, 2014a,b; EPERZAN™, 2014; Epoetin alfa HEXAL®, 2012; EPORATIO®, 2009; ERBITUX®, 2009; ERWINAZE®, 2011; EYLEA®, 2012; EYLEA®, 2013; FABRAZYME®, 2006; FERTAVID®, 2009; FOLLISTIM®, 2011; GAZYVA™, 2014; GAZYVARO®, 2014; Ghaderi et al., 2012; GONAL-F®, 2010; GRANIX™, 2014; HELIXATE® NexGen, 2010; HERCEPTIN®, 2010; HUMIRA®, 2008; HYLENEX®, 2012; ILARIS®, 2014; JETREA®, 2012; JETREA®, 2013; KADCYLA®, 2013, 2014; KOGENATE® Bayer, 2010; KRYSTEXXA®, 2013; LUMIZYME®, 2010; LUVERIS®, 2005; MABTHERA®, 2008; MENVEO®, 2010; METALYSE®, 2006; MYALEPT™, 2014; MYOZYME®, 2011; NAGLAZYME®, 2005; NOVOSEVEN®, 2006; NOVOTHIRTEEN®, 2012; NPLATE®, 2009; NULOJIX®, 2011; OBIZUR™, 2014; Office of Device Evaluation and Center for Devices and Radiological Health, 2001; OPGENRA®, 2014; ORENCIA®, 2012; OVIDREL®, 2014; OVITRELLE®, 2006; PERJETA®, 2013a,b; PREVNAR®, 2009; PROCRIT®, 2000; PROLIA®, 2010; PULMOZYME®, 2010; RAXIBACUMAB™, 2014; REBIF®, 2008; REFACTO AF®, 2014; REMICADE®, 2009; REOPRO®, 2013; RITUXAN®, 2014; ROACTEMRA®, 2013; SAIZEN®, 1987; SEROSTIM®, 1987; SIMPONI®, 2009; SIMULECT®, 2008; Somatropin Biopartners, 2013; STELARA®, 2013; Swiech et al., 2012; SYLVANT®, 2015; SYLVANT™, 2014; SYNAGIS®, 2009; TANZEUM™, 2014; tbo-filgrastim, 2012; THYROGEN®, 2010; TNKASE®, 2011; TRETTEN®, 2014; TYSABRI®, 2011; US Food and Drug Administration, 2010, 2011, 2012, 2013, 2014; Vectibix®, 2014; Victoza®, 2009; VIMIZIM®, 2014a,b; VORAXAZE®, 2012; XEOMIN®, 2014; XIAFLEX®, 2014; XOLAIR®, 2010; XYNTHA®, 2011; YERVOY®, 2011; ZALTRAP®, 2013a,b; ZEVALIN®, 2009).

Bacterial expression systems (e.g. Escherichia coli) possess the advantages of being straightforward to culture, with rapid cell growth and high yields. In addition, protein expression can be initiated through promoter induction by addition of lactose or the lactose analogue isopropyl-β-d-thiogalactopyranoside (IPTG; IPTG induces the promoters lac, tac and trc). However, such systems are unable to produce complex, mammalian-like glycosylation due to the absence of the necessary enzymatic components and the intracellular compartmentalization required (Ghaderi et al., 2012; Graumann & Premstaller, 2006). In addition, mammalian proteins produced in these systems often aggregate, forming inclusion bodies, due to the low solubility of mammalian proteins in prokaryotic cells and absence of appropriate protein chaperone systems. Proteins produced in bacterial expression systems must often be extracted from inclusion bodies and refolded. Bacterial systems are therefore generally used for production of non-glycosylated proteins, including some mAbs, hormones, cytokines and enzymes (Ghaderi et al., 2012; Graumann & Premstaller, 2006).

Similar to bacterial expression systems, yeast expression systems (e.g. Saccharomyces cerevisiae and Pichia pastoris) achieve rapid cell growth and high-protein yields with straightforward production scalability and without the need for animal-derived growth factors (Gerngross, 2004). Yeast cell lines may also be used to produce proteins that cannot be obtained from E. coli due to the problems associated with folding and stereochemistry (Gerngross, 2004). The key challenge associated with yeast expression systems is their production of high mannose residues within their expressed PTMs (50–200 vs three molecules in human cells, as part of either N- or O-linked glycan structures), which may confer a short half-life and render proteins less efficacious and even immunogenic in humans (Dean, 1999; Gemmill & Trimble, 1999; Gerngross, 2004; Lam et al., 2007; Mochizuki et al., 2001). The development of yeasts that have been genetically modified to address the issue of high mannose content has been reported (Chiba et al., 1998; Gerngross, 2004; Ghaderi et al., 2012; Hamilton et al., 2003). The expression of a fully humanized sialylated glycoprotein in glycoengineered yeast constitutes a major advance in the use of yeast expression systems for biopharmaceutical manufacturing (Hamilton & Gerngross, 2007).

Plant and insect cell expression systems are able to produce proteins with complex glycosylation patterns; however, the glycan structures produced are significantly different from those produced in humans (Ghaderi et al., 2012). Plants lack many of the key glycosylated residues present in humans, most notably sialic acids. In addition, they produce α1,3-fructose and β1,2-xylose, which are absent in humans and may be immunogenic (Ghaderi et al., 2012). Notably, in 2012, taliglucerase alfa (ELELYSO®; Pfizer, New York, NY) was approved by the US Food and Drug Administration (FDA) for the treatment of type 1 Gaucher disease. This therapy is produced using genetically modified carrot plant root cells that produce the enzyme with a human compatible glycan profile (ELELYSO™, 2014).

Insect cells infected with the viral vector baculovirus (baculovirus-insect cell expression system) can also efficiently express recombinant proteins, and these systems are mostly used for the development of virus-like particles and, subsequently, vaccines (Kost et al., 2005; Liu et al., 2013). However, although they produce N-glycan precursors, these are trimmed, resulting in either high mannose or paucimannose residues that do not develop further into terminal galactose and/or sialic acid residues (Kost et al., 2005). This is evidenced by the lack of either galactosyltransferase or sialyltransferase activity. As in plants, insect systems may also express the fucosylated α1,3-linkage (Staudacher et al., 1999). However, in recent years, there have been developments in the use of transgenic insect cells, with humanized protein glycosylation mechanisms (Kost et al., 2005).

The majority of currently licensed biotherapeutic products are produced in non-human mammalian expression systems (Table 1), as these systems are able to produce PTMs that (outside of a human expression system) most closely resemble those in humans (Ghaderi et al., 2010). These expression systems are used to produce mAbs, hormones, cytokines, enzymes and clotting factors (Ghaderi et al., 2012).

The most frequently used mammalian system is the CHO cell line, which is used in the manufacture of >70% of currently approved recombinant proteins (Butler & Spearman, 2014). This cell line has demonstrated several major advantages. First, CHO cells are able to grow in suspension culture (which enables large-scale production; other cell lines, such as insect cells, also have this ability) and serum-free chemically defined media (enabling reproducibility across different batches of cultures with a better safety profile than in media that contain human- or animal-derived proteins) (Kim et al., 2012; Lai et al., 2013; Rossi et al., 2012). Historically, CHO cells allowed gene amplification, resulting in a higher recombinant protein yield (up to the gram per liter range for some proteins) and specific productivity, which was previously an issue in other mammalian cell lines (Carlage et al., 2012; Kim et al., 2012; Yang et al., 2014a,b). Other advances, such as the creation of stronger expression units and advanced hosts, better selection strategies (e.g. through technologic advances in screening for high-productivity clones) and targeting the transgene to transcriptional hotspots (site-specific integration of transgenes), also contribute to the high protein yields attained from these cells (Kim et al., 2012). In addition, this expression system is highly tolerant to changes in pH, oxygen level, pressure or temperature during manufacturing (Ghaderi et al., 2012; Lai et al., 2013). Furthermore, due to the long period of time that this cell line has been used, there is a degree of familiarity with the CHO platform within development and manufacturing organizations, regulatory agencies, and suppliers (e.g. cell culture media suppliers), which could potentially decrease overall timelines. This familiarity may also be beneficial when assessing contaminant profiles (e.g. host cell proteins), which may be better characterized for CHO cells compared with newer cell lines.

The first recombinant biotherapeutic protein produced in CHO cells was tissue plasminogen activator, approved in 1986 (Kim et al., 2012). Therefore, the safety profile of CHO cells has been established for more than 20 years (Butler & Spearman, 2014; Kim et al., 2012). CHO cells have been shown to have reduced susceptibility to certain viral infections compared with other mammalian cell lines (e.g. the BHK cell line), and routine screening systems for adventitious agents are effective in detecting cell line infections (Berting et al., 2010). This reduced susceptibility may be due to the fact that many viral entry genes are not expressed in CHO cells (Xu et al., 2011). Further, there is perceived species barrier protection with the use of hamster-derived cells, reducing the potential risk of transfer of contaminating adventitious agents to humans (Berting et al., 2010; Swiech et al., 2012). However, many viruses have the ability to cross the species barrier and may still pose a risk (Pauwels et al., 2007).

Perhaps the most important advantage of CHO cells is that they are able to produce proteins with complex bioactive PTMs that are similar to those produced in humans. However, CHO cells are unable to produce some types of human glycosylation (CHO cells lack α[2-6] sialyltransferase α[1-3/4] fucosyltransferases) and they produce glycans that are not expressed in humans, namely α-gal and NGNA (Bosques et al., 2010; Dietmair et al., 2012; Ghaderi et al., 2012). Circulating antibodies against both of these N-glycans are present in humans, which may lead to increased immunogenicity and altered pharmacokinetics of these products when used in humans (Ghaderi et al., 2010; Padler-Karavani et al., 2008). Additional screening in CHO cells is required in order to isolate clones lacking the α-gal and NGNA glycans. This screening may result in otherwise productive clones needing to be discarded (Ghaderi et al., 2010). However, the attachment of non-human glycans may not be a concern for therapeutic proteins that do not require glycosylation, which illustrates the importance of considering the specific product molecule when choosing an appropriate cell line for production of a protein.

Other mammalian cell lines used for the production of biotherapeutic proteins include BHK-21 cells, used in the production of some coagulation factors such as factor VIII (Wurm, 2004). When murine myeloma cell lines (NS0 and Sp2/0) have been used historically, they have generally been used in the production of mAbs, for example, palivizumab and ofatumumab (Barnes et al., 2000; Butler & Spearman, 2014; Ghaderi et al., 2012). These myeloma cells were derived from immunoglobulin-producing tumor cells that no longer produced their original immunoglobulins; these cells possess the appropriate machinery for producing and secreting these proteins (Barnes et al., 2000).

For proteins produced in all of these non-human cell lines, as well as those produced in human cell lines, potential safety concerns arise from the possibility of process-related contaminants and immunogenicity (World Health Organization, 2013). Process-related contaminants may include infectious agents (viral, bacterial, fungal, etc.) with the potential to result in host infection, nucleic acid contaminants with the potential to integrate into the host genome (theoretical), and other contaminants from the manufacturing process, such as exogenous non-human epitopes (e.g. from animal serum used during the manufacturing process) that can be incorporated into human cells and the resultant biotherapeutic protein (Ghaderi et al., 2012).

Human cell lines used to manufacture licensed products

HEK293 and HT-1080 are the two human cell lines most often used in the production of biotherapeutic proteins, which offer the advantage of producing fully human PTMs (Tables 2 and 3) (Loignon et al., 2008; Swiech et al., 2012).

Table 2. Human cells lines and their therapeutic protein productsa,b.

Cell line Product Indication FDA approval status EMA approval status
HEK293        
  Drotrecogin alfa Severe septicemia/septic shock Approved 2001;  withdrawn 2011 Approved 2002;  withdrawn 2011
  rFVIIIFc Hemophilia A Approved 2014 Submitted 2014
  rFIXFc Hemophilia B Approved 2014 NA
  Dulaglutide Type 2 diabetes Approved 2014 Submitted 2014
  Human-cl rhFVIII Hemophilia A Submitted to the FDA Approved 2014
HT-1080        
  Agalsidase alfa Fabry disease NA Approved 2001
  Epoetin delta Anemia secondary to chronic renal failure NA Approved 2002;  withdrawn 2009 (Europe)
  Idursulfase Hunter syndrome Approved 2006 Approved 2007
  Velaglucerase alfa Type 1 Gaucher disease Approved 2010 Approved 2010
PER.C6        
  CL184 Rabies virus infection Submitted to the FDA NA
  MOR103 Rheumatoid arthritis, multiple sclerosis Phase 1 clinical development Phase 1 clinical development

FDA, US Food and Drug Administration; EMA, European Medicines Agency; HEK, human embryonic kidney; NA, not approved; rFVIIIFc, recombinant factor VIII Fc fusion protein; rFIXFc, recombinant factor IX Fc fusion protein; rhFVIII, recombinant human factor VIII.

aData obtained from publically available resources (October 2014); all approved products may not be included.

bReferences: (ALPROLIX®, 2014; Bakker et al., 2005; Behrens et al., 2014; Casademunt et al., 2012; DYNEPO®, 2007; ELAPRASE®, 2012, 2013; ELOCTATE®, 2014; European Medicines Agency and Committee for Medicinal Products for Human Use, 2014; Glaesner et al., 2010; Octapharma, 2014; REPLAGAL®, 2006; TRULICITY™, 2014; VPRIV®, 2010a,b; XIGRIS®, 2008).

Table 3. Comparison of human cell lines with other expression systems in the production of therapeutic proteins.

Advantages Disadvantages
• Absence of potentially immunogenic PTMs due to  human-compatible glycosylation • Easily grown in suspension serum-free culture • Achieve rapid reproduction • Amenable to a number of transfection methods • Clinical experience is not as extensive as for other cell lines,  although experience is growing • Potential susceptibility to human viral contamination

HEK293 cells are easily grown in suspension serum-free culture, reproduce rapidly, are amenable to a number of transfection methods, and are highly efficient at protein production (Swiech et al., 2012; Thomas & Smart, 2005).

HEK293-H (Berkner, 1993) and 293-F (Vink et al., 2014) cell lines are clonal isolates of the HEK293 cell line that were selected for fast growth in serum-free medium, superior transfection efficiency, and a high level of protein production (Gibco, 2014). Subclone 293-H also has improved adherence to monolayer culture (when serum-supplemented media are used) compared with other cell lines. Other modified HEK293 cells include the HEK293-T cell line and HEK293-EBNA1 cells. The HEK293-T (293-T) cell line expresses the simian virus 40 large T antigen and is capable of expressing high titers of viral gene vectors for use in gene therapy (Yamaguchi et al., 2003). HEK293-T cells are often used for the production of retroviral vectors (Yamaguchi et al., 2003). HEK293-EBNA1 cells stably express the Epstein-Barr virus EBNA-1 gene, controlled by the cytomegalovirus promoter and demonstrate a greater growth rate and maximal cell density relative to parental HEK293 cells (Schlaeger & Christensen, 1999).

HEK293 cells have been widely used to produce research-grade proteins for many years and, more recently, five therapeutic agents produced in HEK293 cells have been approved by the FDA or the European Medicines Agency (EMA) for therapeutic use. These agents are drotrecogin alfa (XIGRIS®; Eli Lilly Corporation, Indianapolis, IN), recombinant factor IX Fc fusion protein (rFIXFc; Biogen, Cambridge, MA), recombinant factor VIII Fc fusion protein (rFVIIIFc; Biogen, Cambridge, MA), human cell line recombinant factor VIII (human-cl rhFVIII; NUWIQ®; Octapharma, Lachen, Switzerland) and dulaglutide (TRULICITY®; Eli Lily, Indianapolis, IN).

Drotrecogin alfa is a recombinant activated protein C that was approved by the FDA in 2001 and by the EMA in 2002 for the treatment of patients with severe sepsis. HEK293 cells were chosen by the manufacturer for production of drotrecogin alfa because its activity required two PTMs, propeptide cleavage and γ-carboxylation of its glutamic acid residues, which CHO cells cannot produce with adequate efficiency (Berkner, 1993; Durocher & Butler, 2009). The product was approved (Bernard et al., 2001), but was later voluntarily withdrawn from the market by its manufacturer (Eli Lilly) in 2011 following the randomized placebo-controlled Prospective Recombinant Human Activated Protein C Worldwide Evaluation in Severe Sepsis and Septic Shock (PROWESS-SHOCK) trial, which demonstrated no mortality benefit with drotrecogin alfa compared with placebo for patients experiencing septic shock (Green et al., 2012; Ranieri et al., 2012).

rFVIIIFc and rFIXFc are recombinant fusion proteins that were approved by the FDA in 2014 for the control and prevention of bleeding episodes, perioperative management and routine prophylaxis to prevent or reduce the frequency of bleeding episodes in people with hemophilia A and B, respectively (ALPROLIX®, 2014; ELOCTATE®, 2014; Mahlangu et al., 2014; Powell et al., 2013). They are also approved in Canada, Australia and Japan. rFVIIIFc consists of B domain–deleted recombinant factor VIII genetically fused to the Fc portion of immunoglobulin G1 (IgG1) and is produced in HEK293-H cells (Dumont et al., 2012; ELOCTATE®, 2014; Peters et al., 2013). The rFVIIIFc fed-batch culture process is robust at scales of 200, 2000 and 15 000 liters, with the potential for a second-generation process to achieve even higher cell densities, on the order of 3.5 × 107 vc/ml (Huang et al., 2014). rFIXFc was also produced using HEK293-H cells, and consists of the factor IX sequence covalently linked to the Fc domain of human IgG1 (ALPROLIX®, 2014; Durocher & Butler, 2009; McCue et al., 2014; Peters et al., 2010). An essential PTM for FIX activity is γ-carboxylation of the first 12 glutamic acid residues in the Gla domain by vitamin K–dependent γ-glutamyl carboxylase. This modification facilitates binding of FIX to phospholipid membranes. HEK293 cells have been reported to have a greater capacity for γ-carboxylation than CHO cells (Berkner, 1993). Furthermore, FVIII contains six potential tyrosine sulfation sites, which are vital for FVIII functionality and binding to von Willebrand factor. FVIII expressed from human cell lines has been reported to be fully sulfated (Kannicht et al., 2013; Peters et al., 2013).

The use of a human cell line for replacement coagulation factors, such as rFVIIIFc and rFIXFc, may result in reduced immunogenicity relative to non-human mammalian cell lines, as α-gal and NGNA glycan moieties are absent from these manufactured protein products (Bosques et al., 2010; McCue et al., 2014, 2015; Noguchi et al., 1995). However, it should be noted that several recombinant clotting factor products produced in non-human mammalian cell lines have been used successfully for many years. The development of inhibitors (neutralizing antibodies) against replacement clotting factors occurs in ∼30% of people with severe hemophilia A and 5% of those with severe hemophilia B. The causative F8 or F9 gene mutation plays a pivotal role in inhibitor development in hemophilia A and B, respectively, with large or complete deletions, non-sense mutations or inversions (e.g. intron 22 inversion in the F8 gene) being the most commonly associated mutations (Franchini & Mannucci, 2011). The impact of PTMs on inhibitor development is unknown, and will need further research. Importantly, none of the previously treated people with hemophilia in the phase 1/2a or phase 3 clinical studies developed inhibitors to the rFVIIIFc and rFIXFc fusion products (Mahlangu et al., 2014; Powell et al., 2012, 2013; Shapiro et al., 2012).

Human-cl rhFVIII (NUWIQ®), an additional factor VIII replacement product for the management of hemophilia A, is being produced in the HEK293-F cell line. Like HEK293-H cells, HEK293-F cells are a derivation of HEK293 cells that have been pre-adapted for growth in serum-free culture medium (Casademunt et al., 2012). Human-cl rhFVIII has been approved by the EMA and submitted to the FDA for approval (Octapharma, 2014); this product has been shown to exhibit a similar glycosylation profile to human plasma-derived factor VIII, without α-gal and NGNA (Kannicht et al., 2013).

Glucagon-1-like peptide (GLP-1) Fc fusion protein (dulaglutide) has been approved by the FDA for the treatment of type 2 diabetes mellitus, and is produced using HEK293-EBNA cells (Glaesner et al., 2010; TRULICITY™, 2014). Large clinical trials have demonstrated its superiority over the dipeptidyl peptidase-4 inhibitor antagonist exenatide and its non-inferiority to liraglutide (a GLP-1 agonist), when added on to oral diabetic agents (Dungan et al., 2014; Wysham et al., 2014).

Another human cell line, HT-1080, was produced from a fibrosarcoma with an epithelial-like phenotype (Swiech et al., 2012). With the use of gene activation technology (in which the endogenous DNA promoter is replaced with a more potent type), four approved therapeutic proteins have been produced by Shire (Swiech et al., 2012).

1) Epoetin delta (DYNEPO®) was approved by the EMA in 2002 for the treatment of anemia secondary to chronic renal failure (DYNEPO®, 2007; ELAPRASE®, 2013; REPLAGAL®, 2006; Swiech et al., 2012; VPRIV®, 2013). However, this has been voluntarily withdrawn by the manufacturer for commercial reasons.

2) Iduronate-2-sulfatase (idursulfase; ELAPRASE®) is licensed as enzyme replacement therapy (EMA in 2007 and FDA in 2006) for the treatment of Hunter syndrome (mucopolysaccharidosis II), an X-linked lysosomal storage disorder (ELAPRASE®, 2013).

3) Agalsidase alfa (REPLAGAL®; Shire Human Genetic Therapies, Danderyd, Sweden) was approved by the EMA in 2001 for the treatment of Fabry disease (REPLAGAL®, 2010). Compared with agalsidase beta (FABRAZYME®; Genzyme Therapeutics, Cambridge, MA), which is produced using CHO cells for a similar indication (FABRAZYME®, 2010, 2014), agalsidase alfa has shown similar enzyme kinetics. However, agalsidase alfa demonstrates a lesser uptake into fibroblasts from patients with Fabry disease and also lower concentrations in the kidney, heart and spleen of mice (Lee et al., 2003). A single clinical study has compared the two products; this showed no significant differences for all efficacy outcomes, and there were no differences for the development of antibodies (Vedder et al., 2007).

4) The fourth agent produced in HT-1080 cells, velaglucerase alfa (VPRIV®; Shire Human Genetic Therapies, Lexington, MA), was approved in 2010 (FDA and EMA) for the treatment of type 1 Gaucher disease (DYNEPO®, 2007; ELAPRASE®, 2013; REPLAGAL®, 2006; Swiech et al., 2012; VPRIV®, 2013). Velaglucerase alfa has been compared with two similar products: imiglucerase, produced using CHO cells, and taliglucerase alfa, produced using carrot cells (Ben Turkia et al., 2013; Tekoah et al., 2013).

These products have diverse glycan profiles and the studies have generally shown comparable uptake into macrophages, in vitro enzymatic activity, stability, organ distribution and efficacy (Ben Turkia et al., 2013; Tekoah et al., 2013). However, neutralizing antibodies to imiglucerase were noted in 24% of patients, which had an impact on enzyme activity. It was noted that various factors, such as the production cell line and glycosylation, may be responsible for the difference in immunogenicity, however, the specificity of the anti-imiglucerase antibodies was not stated (Ben Turkia et al., 2013).

Notably, studies that evaluated epoetin delta produced in HT-1080 cells demonstrated differences in glycosylation compared with erythropoietin produced in CHO cells, including a lack of NGNA in the proteins (Butler & Spearman, 2014; Llop et al., 2008; Shahrokh et al., 2011). However, there were additional overlapping isoforms present in endogenous human erythropoietin isolated from urine and serum relative to epoetin delta that could not be accounted for by sialic residues alone.

Human cell lines used in the expression of proteins in clinical and preclinical development

Human cell lines have been extensively utilized for the production of products that are currently in clinical development. In addition, human cell lines are a frequently used expression system for biomedical research due to their production of human PTMs and high productivity. As productivity may vary across clonal isolates, it is important to screen for those clones with the highest yield of the therapeutic protein (Berkner, 1993).

The PER.C6 cell line was created from human embryonic retinal cells, immortalized via transfection with the adenovirus E1 gene (Havenga et al., 2008). This system was originally developed for the production of human adenovirus vectors for use in vaccine development and gene therapy (Butler & Spearman, 2014). An investment was made in this cell line in order to develop a human expression system, and now an advantage of PER.C6 is its ability to produce a high level of protein when used in the production of human IgG (Jones et al., 2003). However, this does not require amplification of the incorporated gene (Jones et al., 2003). Currently, a variety of products utilizing the PER.C6 cell line are in phase 1 or 2 clinical trials (Durocher & Butler, 2009), including the MOR103 mAb, a human IgG antibody against granulocyte macrophage colony-stimulating factor, and CL184, an antibody against the rabies virus (Nagarajan et al., 2014).

MOR103 is in clinical development for the treatment of rheumatoid arthritis and multiple sclerosis. In a phase 1b/2a, randomized, placebo-controlled study, MOR103 was active in patients with moderately severe rheumatoid arthritis; a small number of patients developed anti-MOR103 antibodies (Behrens et al., 2014). CL184 is a combination of two mAbs, human IgG1(λ) and human IgG1(κ) (Bakker et al., 2005). In a phase 1 clinical study, it demonstrated a favorable safety profile and rapid development of rabies virus neutralizing activity, while there was no evidence to suggest the development of human anti-human antibodies (Bakker et al., 2008). CL184 has been granted FDA fast-track approval status.

Two additional cell lines are utilized by products currently in preclinical development. The CAP cell line is derived from human amniocytes obtained through amniocentesis; these cells are immortalized through an adenovirus type 5 E1 gene (Schiedner et al., 2008; Swiech et al., 2011). In addition to the ability to produce human PTMs, the primary advantage of this cell is the potential for high protein yields (Schiedner et al., 2008).

The HKB-11 cell line was created through polyethylene glycol fusion of HEK293-S and a human B-cell line (modified Burkitt lymphoma cells) (Cho et al., 2003; Durocher & Butler, 2009; Picanco-Castro et al., 2013). The advantages of this cell line include high-level protein production without the formation of aggregates, which can be a problem in other human cell lines (Picanco-Castro et al., 2013). Notably, HKB-11 has demonstrated increased expression of human FVIII compared with expression in HEK293 and BHK21 (Mei et al., 2006). Similar to other human cell lines, it has been shown to produce human glycosylation patterns including α (2,3) and α (2,6) sialic acid linkages (Picanco-Castro et al., 2013). HKB-11 has been used to produce a recombinant factor VIII protein and tissue factor (Cho et al., 2003).

A more recently developed cell line, HuH-7, originates from a human hepatocellular carcinoma (Enjolras et al., 2012). A recent study has shown that the HuH-7-CD4 clone is capable of producing recombinant human factor IX with a human glycosylation profile. PTM profiles (e.g. glycosylation, sialylation, phosphorylation and sulfation) were similar to plasma-derived and recombinant factor IX (rFIX), and were improved relative to rFIX produced in CHO cells (Enjolras et al., 2012). More recently, the HuH-7 cell line has been used to produce mutant forms of rFIX that have improved binding affinity for activated FVIII, and also demonstrated enhanced clotting activity in mice (Perot et al., 2015).

Perceptions of risks versus benefits of using human cell lines

The human-specific glycosylation pattern of the PTMs produced by human cell lines offer several advantages compared with those produced in animal cell lines. Although other mammalian cells can produce similar PTMs to human cells, most also produce α-gal and NGNA, PTMs that are not present in the structure of human proteins (Ghaderi et al., 2012). Patterns of post-translational glycosylation are known to affect protein yield, bioactivity, and clearance (Ghaderi et al., 2010). In addition, antibodies to NGNA have been widely reported to occur in humans (Chung et al., 2008; Ghaderi et al., 2012). One study utilizing an NGNA knockout mouse model demonstrated increased immunogenicity of cetuximab due to anti-NGNA antibodies (Ghaderi et al., 2010). In addition, in patients receiving the mAb cetuximab for the treatment of colorectal or head and neck cancers, the majority of severe hypersensitivity reactions observed in clinical trials were associated with pre-existing IgE antibodies against α-gal (Chung et al., 2008; Ghaderi et al., 2012). Such antibodies may alter the efficacy or immunogenicity of proteins with the presence of non-human glycan structures. Thus, human cell lines can serve as a valuable niche expression system for biotherapeutic proteins that require human PTMs. A theoretical concern with the use of human cell lines is an increased risk of transfer of human adventitious agents, given the lack of a species barrier (Swiech et al., 2012). However, current manufacturing technologies, typically inclusive of multiple viral inactivation or clearance steps, such as nanofiltration, have largely mitigated this concern and may provide more effective viral clearance than has been observed in CHO cells (Kelley et al., 2010; McCue et al., 2014, 2015).

Future perspectives

Production of biotherapeutic proteins in human cell lines is expanding, with several products currently approved for clinical use and others in clinical development in different therapeutic areas. Advantages of human expression systems include achieving equal productivity to other mammalian cell lines and the production of proteins that lack potentially immunogenic, non-human PTMs (most notably α-gal and NGNA). In the future, with additional research investments and a continuation of the technologic advances that have already led to improvements in the use of human cell lines for protein manufacture, human cell lines will be further optimized, more sophisticated product collection strategies will be developed, and these cell lines may become one of the preferred platforms for protein biotherapeutic production.

Acknowledgement

All brand names are trademarks of their respective owners.

Declaration of interest

Editorial support for the writing of this manuscript was provided by Melissa Yuan, MD, of MedErgy, and was funded by Biogen. All authors are employees of and hold equity interest in Biogen.

References

  1. ABSEAMED® . (1,000 IU/0.5 mL solution for injection in a pre-filled syringe) [summary of product characteristics] Kundl, Austria: Sandoz GmbH; 2012. [Google Scholar]
  2. ACTEMRA® . (Tocilizumab) injection, for intravenous use injection, for subcutaneous use [package insert] South San Francisco, CA: Genentech, Inc; 2013. [Google Scholar]
  3. ACTILYSE® . (Alteplase, recombinant tissue plasminogen activator, rt-PA) [package insert] North Ryde, North South Wales, Australia: Boehringer Ingelheim Pty Limited; 2014. [Google Scholar]
  4. ACTIVASE® . (Alteplase) a recombinant tissue plasminogen activator [package insert] South San Francisco, CA: Genentech, Inc; 2012. [Google Scholar]
  5. ADCETRIS® . ([Brentuximab vedotin] for injection) [package insert] Bothell, WA: Seattle Genetics, Inc; 2013. [Google Scholar]
  6. ADVATE® . (250 IU powder and solvent for solution for injection) [summary of product characteristics] Vienna, Austria: Baxter AG; 2014. [Google Scholar]
  7. ALDURAZYME® . (100 U/mL concentrate for solution for infusion) [summary of product characteristics] Haverhill, Suffolk: Genzyme Ltd; 2008. [Google Scholar]
  8. ALPROLIX® . (Coagulation factor IX [recombinant] Fc fusion protein) [package insert] Cambridge, MA: Biogen Idec, Inc; 2014. [Google Scholar]
  9. ARANESP® . (10 micrograms solution for injection in pre-filled syringe) [summary of product characteristics] Breda, The Netherlands: Amgen Europe B.V; 2006. [Google Scholar]
  10. ARCALYST™ . (Rilonacept) injection for subcutaneous use [package insert] Tarrytown, NY: Regeneron Pharmaceuticals, Inc; 2008. [Google Scholar]
  11. Arnold JN, Wormald MR, Sim RB, et al. The impact of glycosylation on the biological function and structure of human immunoglobulins. Annu Rev Immunol. 2007;25:21–50. doi: 10.1146/annurev.immunol.25.022106.141702. [DOI] [PubMed] [Google Scholar]
  12. ARZERRA® . (100 mg concentrate for solution for infusion) [summary of product characteristics] Brentford, Middlesex: Glaxo Group Ltd; 2014. [Google Scholar]
  13. AVASTIN® . (25 mg/mL concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2010. [Google Scholar]
  14. AVONEX® . (30 micrograms powder and solvent for solution for injection) [summary of product characteristics] Maidenhead, Berkshire: Biogen Idec Limited; 2007. [Google Scholar]
  15. Bakker AB, Marissen WE, Kramer RA, et al. Novel human monoclonal antibody combination effectively neutralizing natural rabies virus variants and individual in vitro escape mutants. J Virol. 2005;79:9062–8. doi: 10.1128/JVI.79.14.9062-9068.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bakker AB, Python C, Kissling CJ, et al. First administration to humans of a monoclonal antibody cocktail against rabies virus: safety, tolerability, and neutralizing activity. Vaccine. 2008;26:5922–7. doi: 10.1016/j.vaccine.2008.08.050. [DOI] [PubMed] [Google Scholar]
  17. Barnes LM, Bentley CM, Dickson AJ. Advances in animal cell recombinant protein production: GS-NS0 expression system. Cytotechnology. 2000;32:109–23. doi: 10.1023/A:1008170710003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Beck M. Agalsidase alfa for the treatment of Fabry disease: new data on clinical efficacy and safety. Expert Opin Biol Ther. 2009;9:255–61. doi: 10.1517/14712590802658428. [DOI] [PubMed] [Google Scholar]
  19. Behrens F, Tak PP, Ostergaard M, et al. MOR103, a human monoclonal antibody to granulocyte-macrophage colony-stimulating factor, in the treatment of patients with moderate rheumatoid arthritis: results of a phase Ib/IIa randomised, double-blind, placebo-controlled, dose-escalation trial. Ann Rheum Dis. 2015;74:1058–64. doi: 10.1136/annrheumdis-2013-204816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ben Turkia H, Gonzalez DE, Barton NW, et al. Velaglucerase alfa enzyme replacement therapy compared with imiglucerase in patients with Gaucher disease. Am J Hematol. 2013;88:179–84. doi: 10.1002/ajh.23382. [DOI] [PubMed] [Google Scholar]
  21. BENEFIX® . (250 IU powder and solvent for solution for injection) [summary of product characteristics] Sandwich, Kent: Pfizer Limited; 2012. [Google Scholar]
  22. BENLYSTA® . (120 mg powder for concentrate for solution for infusion) [summary of product characteristics] Brentford, Middlesex: Glaxo Group Limited; 2011. [Google Scholar]
  23. Berkner KL. Expression of recombinant vitamin K-dependent proteins in mammalian cells: factors IX and VII. Methods Enzymol. 1993;222:450–77. doi: 10.1016/0076-6879(93)22029-f. [DOI] [PubMed] [Google Scholar]
  24. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001;344:699–709. doi: 10.1056/NEJM200103083441001. [DOI] [PubMed] [Google Scholar]
  25. Berting A, Farcet MR, Kreil TR. Virus susceptibility of Chinese hamster ovary (CHO) cells and detection of viral contaminations by adventitious agent testing. Biotechnol Bioeng. 2010;106:598–607. doi: 10.1002/bit.22723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bosques CJ, Collins BE, Meador JW, III, et al. Chinese hamster ovary cells can produce galactose-alpha-1,3-galactose antigens on proteins. Nat Biotechnol. 2010;28:1153–6. doi: 10.1038/nbt1110-1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Butler M, Spearman M. The choice of mammalian cell host and possibilities for glycosylation engineering. Curr Opin Biotechnol. 2014;30C:107–12. doi: 10.1016/j.copbio.2014.06.010. [DOI] [PubMed] [Google Scholar]
  28. CAMPATH® . (Alemtuzumab) injection for intravenous use [package insert] Cambridge, MA: Genzyme Corporation; 2014. [Google Scholar]
  29. Carlage T, Kshirsagar R, Zang L, et al. Analysis of dynamic changes in the proteome of a Bcl-XL overexpressing Chinese hamster ovary cell culture during exponential and stationary phases. Biotechnol Prog. 2012;28:814–23. doi: 10.1002/btpr.1534. [DOI] [PubMed] [Google Scholar]
  30. Casademunt E, Martinelle K, Jernberg M, et al. The first recombinant human coagulation factor VIII of human origin: human cell line and manufacturing characteristics. Eur J Haematol. 2012;89:165–76. doi: 10.1111/j.1600-0609.2012.01804.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. CATHFLO® ACTIVASE® . (Alteplase) powder for reconstitution for use in central venous access devices [package insert] South San Francisco, CA: Genentech, Inc; 2010. [Google Scholar]
  32. CEREZYME® . (200 U powder for concentrate for solution for infusion) [summary of product characteristics] Naarden, The Netherlands: Genzyme Europe B.V; 2010. [Google Scholar]
  33. CERVARIX® . (Suspension for injection; human papillomavirus vaccine [types 16, 18; recombinant, adjuvanted, adsorbed]) [summary of product characteristics] Rixensart, Belgium: GlaxoSmithKline Biologicals s.a; 2012. [Google Scholar]
  34. Chiba Y, Suzuki M, Yoshida S, et al. Production of human compatible high mannose-type (Man5GlcNAc2) sugar chains in Saccharomyces cerevisiae . J Biol Chem. 1998;273:26298–304. doi: 10.1074/jbc.273.41.26298. [DOI] [PubMed] [Google Scholar]
  35. Cho MS, Yee H, Brown C, et al. Versatile expression system for rapid and stable production of recombinant proteins. Biotechnol Prog. 2003;19:229–32. doi: 10.1021/bp0255964. [DOI] [PubMed] [Google Scholar]
  36. Chung CH, Mirakhur B, Chan E, et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N Engl J Med. 2008;358:1109–17. doi: 10.1056/NEJMoa074943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. CIMZIA® . (Certolizumab pegol) for injection, for subcutaneous use [package insert] Smyrna, GA: UCB, Inc; 2013. [Google Scholar]
  38. CIMZIA®. (2014). Cimzia 200 mg solution for injection [summary of product characteristics]. Bruxelles, Belgium: UCB Pharma S.A [Google Scholar]
  39. CYRAMZA® . ([Ramucirumab] injection, for intravenous infusion) [package insert] Indianapolis, IN: Eli Lilly and Company; 2014. [Google Scholar]
  40. Dean N. Asparagine-linked glycosylation in the yeast Golgi. Biochim Biophys Acta. 1999;1426:309–22. doi: 10.1016/s0304-4165(98)00132-9. [DOI] [PubMed] [Google Scholar]
  41. Dietmair S, Hodson MP, Quek LE, et al. A multi-omics analysis of recombinant protein production in Hek293 cells. PLoS One. 2012;7:e43394. doi: 10.1371/journal.pone.0043394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Dumont JA, Liu T, Low SC, et al. Prolonged activity of a recombinant factor VIII-Fc fusion protein in hemophilia A mice and dogs. Blood. 2012;119:3024–30. doi: 10.1182/blood-2011-08-367813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Dungan KM, Povedano ST, Forst T, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet. 2014;384:1349–57. doi: 10.1016/S0140-6736(14)60976-4. [DOI] [PubMed] [Google Scholar]
  44. Durocher Y, Butler M. Expression systems for therapeutic glycoprotein production. Curr Opin Biotechnol. 2009;20:700–7. doi: 10.1016/j.copbio.2009.10.008. [DOI] [PubMed] [Google Scholar]
  45. DYNEPO® . (1,000 IU/0.5 mL solution for injection in a pre-filled syringe) [summary of product characteristics] Basingstoke, Hampshire: Shire Pharmaceutical Contracts Ltd; 2007. [Google Scholar]
  46. ELAPRASE® . (2 mg/mL concentrate for solution for infusion) [summary of product characteristics] Danderyd, Sweden: Shire Human Genetic Therapies AB; 2012. [Google Scholar]
  47. ELAPRASE® . ([Idursulfase] injection, for intravenous use) [package insert]. Lexington, MA: Shire Human Genetic Therapies, Inc; 2013. [Google Scholar]
  48. ELELYSO™ . ([Taliglucerase alfa] for injection, for intravenous use) [package insert] New York, NY: Shire Human Genetic Therapies, Inc; 2014. [Google Scholar]
  49. ELOCTATE® . ([Antihemophilic factor (recombinant) Fc fusion protein] lyophilized powder for solution for intravenous injection) [package insert] Cambridge, MA: Biogen Idec Inc; 2014. [Google Scholar]
  50. ENBREL® . (25 mg powder and solvent for solution for injection) [summary of product characteristics] Sandwich, Kent: Pfizer Limited; 2010. [Google Scholar]
  51. Enjolras N, Dargaud Y, Perot E, et al. Human hepatoma cell line HuH-7 is an effective cellular system to produce recombinant factor IX with improved post-translational modifications. Thromb Res. 2012;130:e266–73. doi: 10.1016/j.thromres.2012.08.313. [DOI] [PubMed] [Google Scholar]
  52. ENTYVIO® . ([Vedolizumab] for injection, for intravenous use) [package insert] Deerfield, IL: Takeda Pharmaceuticals America Inc; 2014a. [Google Scholar]
  53. ENTYVIO® . (300 mg powder for concentrate for solution for infusion) [summary of product characteristics] Taastrup, Denmark: Takeda Pharma A/S; 2014b. [Google Scholar]
  54. EPERZAN™ . (30 mg powder and solvent for solution for injection) [summary of product characteristics] Cork, Ireland: GlaxoSmithKline Trading Services Limited; 2014. [Google Scholar]
  55. Epoetin alfa HEXAL® . (1,000 IU/0.5 mL solution for injection in a pre-filled syringe) [summary of product characteristics] Holzkirchen, Germany: Hexal AG; 2012. [Google Scholar]
  56. EPORATIO® . (1,000 IU/0.5 mL Solution for injection in pre-filled syringe) [summary of product characteristics] Ulm, Germany: ratiopharm GmbH; 2009. [Google Scholar]
  57. ERBITUX® . (5 mg/mL solution for infusion) [summary of product characteristics] Darmstadt, Germany: Merck KGaA; 2009. [Google Scholar]
  58. ERWINAZE® . ([Asparaginase Erwinia chrysanthemi] for injection, intramuscular use) [package insert] Langhorne, PA: EUSA Pharma (USA), Inc; 2011. [Google Scholar]
  59. Estes S, Melville M. Mammalian cell line developments in speed and efficiency. Adv Biochem Eng Biotechnol. 2014;139:11–33. doi: 10.1007/10_2013_260. [DOI] [PubMed] [Google Scholar]
  60. European Medicines Agency, Committee for Medicinal Products for Human Use. (2014). Summary of opinion (initial authorisation) Trulicity dulaglutide. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Summary_of_opinion_-_Initial_authorisation/human/002825/WC500173650.pdf. [last accessed 8 Dec 2014]
  61. EYLEA® . (40 mg/mL solution for injection in pre-filled syringe) [summary of product characteristics] Berlin, Germany: Bayer Pharma AG; 2012. [Google Scholar]
  62. EYLEA® . (40 mg/mL solution for injection in a vial) [summary of product characteristics] Newbury, Berkshire: Bayer plc; 2013. [Google Scholar]
  63. FABRAZYME® . (35 mg powder for concentrate for solution for infusion) [summary of product characteristics] Naarden, The Netherlands: Genzyme Europe B.V; 2006. [Google Scholar]
  64. FABRAZYME®. (2014). Fabrazyme. European Product Assessment Report. Available from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000370/human_med_000784.jsp&mid=WC0b01ac058001d124. [last accessed 8 Dec 2014]
  65. FABRAZYME® . ([Agalsidase beta] injection, powder, lyophilized for solution for intravenous use) [package insert] Cambridge, MA: Genzyme Corporation; 2010. [Google Scholar]
  66. FERTAVID® . (50 IU/0.5 mL solution for injection) [summary of product characteristics] Hoddesdon, Hertfordshire: Merck, Sharp & Dohme Ltd; 2009. [Google Scholar]
  67. FOLLISTIM® . AQ Cartridge (follitropin beta injection) for subcutaneous use [package insert] Whitehouse Station, NJ: Merck Sharp & Dohme B.V., a subsidiary of Merck & Co., Inc; 2011. [Google Scholar]
  68. Franchini M, Mannucci PM. Inhibitors of propagation of coagulation (factors VIII, IX and XI): a review of current therapeutic practice. Br J Clin Pharmacol. 2011;72:553–62. doi: 10.1111/j.1365-2125.2010.03899.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. GAZYVA™ . ([Obinutuzumab] injection, for intravenous infusion) [package insert] South San Francisco: Genentech, Inc; 2014. [Google Scholar]
  70. GAZYVARO® . Gazyvaro (1,000 mg concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2014. [Google Scholar]
  71. Gemmill TR, Trimble RB. Overview of N- and O-linked oligosaccharide structures found in various yeast species. Biochim Biophys Acta. 1999;1426:227–37. doi: 10.1016/s0304-4165(98)00126-3. [DOI] [PubMed] [Google Scholar]
  72. Gerngross TU. Advances in the production of human therapeutic proteins in yeasts and filamentous fungi. Nat Biotechnol. 2004;22:1409–14. doi: 10.1038/nbt1028. [DOI] [PubMed] [Google Scholar]
  73. Ghaderi D, Taylor RE, Padler-Karavani V, et al. Implications of the presence of N-glycolylneuraminic acid in recombinant therapeutic glycoproteins. Nat Biotechnol. 2010;28:863–7. doi: 10.1038/nbt.1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Ghaderi D, Zhang M, Hurtado-Ziola N, et al. Production platforms for biotherapeutic glycoproteins. Occurrence, impact, and challenges of non-human sialylation. Biotechnol Genet Eng Rev. 2012;28:147–75. doi: 10.5661/bger-28-147. [DOI] [PubMed] [Google Scholar]
  75. Gibco. (2014). Gibco 293-F cells, SFM adapted. Grand Island, NY: Life Technologies [online]. Available from: http://www.lifetechnologies.com/order/catalog/product/11625019?ICID=search-product
  76. Glaesner W, Vick AM, Millican R, et al. Engineering and characterization of the long-acting glucagon-like peptide-1 analogue LY2189265, an Fc fusion protein. Diabetes Metab Res Rev. 2010;26:287–96. doi: 10.1002/dmrr.1080. [DOI] [PubMed] [Google Scholar]
  77. GONAL-F® . (75 IU [5.5 micrograms] powder and solvent for solution for injection) [summary of product characteristics] London, UK: Merck Serono Europe Ltd; 2010. [Google Scholar]
  78. GRANIX™ . (tbo-filgrastim) Injection, for subcutaneous use [package insert] North Wales, PA: Teva Pharmaceuticals USA, Inc; 2014. [Google Scholar]
  79. Graumann K, Premstaller A. Manufacturing of recombinant therapeutic proteins in microbial systems. Biotechnol J. 2006;1:164–86. [Google Scholar]
  80. Green RS, Djogovic D, Howes D. Sepsis update: management of severe sepsis and septic shock in the emergency department after the withdrawal of Xigris. CJEM. 2012;14:265–9. doi: 10.2310/8000.2012.120769. [DOI] [PubMed] [Google Scholar]
  81. Hamilton SR, Bobrowicz P, Bobrowicz B, et al. Production of complex human glycoproteins in yeast. Science. 2003;301:1244–6. doi: 10.1126/science.1088166. [DOI] [PubMed] [Google Scholar]
  82. Hamilton SR, Gerngross TU. Glycosylation engineering in yeast: the advent of fully humanized yeast. Curr Opin Biotechnol. 2007;18:387–92. doi: 10.1016/j.copbio.2007.09.001. [DOI] [PubMed] [Google Scholar]
  83. Havenga MJ, Holterman L, Melis I, et al. Serum-free transient protein production system based on adenoviral vector and PER.C6 technology: high yield and preserved bioactivity. Biotechnol Bioeng. 2008;100:273–83. doi: 10.1002/bit.21757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. HELIXATE® . NexGen (250 IU powder and solvent for solution for injection) [summary of product characteristics] Berlin, Germany: Bayer Pharma AG; 2010. [Google Scholar]
  85. HERCEPTIN® . (150 mg powder for concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2010. [Google Scholar]
  86. Ho RJ, Chien J. Trends in translational medicine and drug targeting and delivery: new insights on an old concept-targeted drug delivery with antibody-drug conjugates for cancers. J Pharm Sci. 2014;103:71–7. doi: 10.1002/jps.23761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Huang YM, Kshirsagar R, Woppman B, et al. In: Cell culture based production. Therapeutic Fc-proteins. Chamow SM, Ryll T, Lowman HB, et al., editors. Weinheim, Germany: Wiley-VCH Verglag GmbH & Co.; 2014. pp. 67–96. [Google Scholar]
  88. HUMIRA® . (40 mg/0.8 mL solution for injection for paediatric use) [summary of product characteristics] Maidenhead, UK: AbbVie Ltd; 2008. [Google Scholar]
  89. HYLENEX® . Recombinant (hyaluronidase human injection) [package insert] San Diego, CA: Halozyme Therapeutics, Inc; 2012. [Google Scholar]
  90. ILARIS® . (150 mg powder for solution for injection) [summary of product characteristics] Horsham, West Sussex: Novartis Europharm Limited; 2014. [Google Scholar]
  91. JETREA® . ([ocriplasmin] intravitreal injection, 2.5 mg/mL) [package insert] Iselin, NJ: ThromboGenics, Inc; 2012. [Google Scholar]
  92. JETREA® . JETREA (0.5 mg/0.2 mL concentrate for solution for injection) [summary of product characteristics] Leuven, Belgium: ThromboGenics NV; 2013. [Google Scholar]
  93. Jones D, Kroos N, Anema R, et al. High-level expression of recombinant IgG in the human cell line per.c6. Biotechnol Prog. 2003;19:163–8. doi: 10.1021/bp025574h. [DOI] [PubMed] [Google Scholar]
  94. KADCYLA® . ([ado-Trastuzumab emtansine] for injection, for intravenous use) [package insert] South San Francisco, CA: Genentech Inc; 2014. [Google Scholar]
  95. KADCYLA® . (100 mg powder for concentrate for solution for infusion; 160 mg powder for concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2013. [Google Scholar]
  96. Kannicht C, Ramstrom M, Kohla G, et al. Characterisation of the post-translational modifications of a novel, human cell line-derived recombinant human factor VIII. Thromb Res. 2013;131:78–88. doi: 10.1016/j.thromres.2012.09.011. [DOI] [PubMed] [Google Scholar]
  97. Kelley B, Jankowski M, Booth J. An improved manufacturing process for Xyntha/ReFacto AF. Haemophilia. 2010;16:717–25. doi: 10.1111/j.1365-2516.2009.02160.x. [DOI] [PubMed] [Google Scholar]
  98. Kim JY, Kim YG, Lee GM. CHO cells in biotechnology for production of recombinant proteins: current state and further potential. Appl Microbiol Biotechnol. 2012;93:917–30. doi: 10.1007/s00253-011-3758-5. [DOI] [PubMed] [Google Scholar]
  99. KOGENATE® . Bayer (250 IU powder and solvent for solution for injection) [summary of product characteristics] Berlin, Germany: Bayer Pharma AG; 2010. [Google Scholar]
  100. Kost TA, Condreay JP, Jarvis DL. Baculovirus as versatile vectors for protein expression in insect and mammalian cells. Nat Biotechnol. 2005;23:567–75. doi: 10.1038/nbt1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. KRYSTEXXA® . (8 mg concentrate for solution for infusion) [summary of product characteristics] Dublin, Ireland: Savient Pharma Ireland Limited; 2013. [Google Scholar]
  102. Lai T, Yang Y, Ng SK. Advances in mammalian cell line development technologies for recombinant protein production. Pharmaceuticals (Basel) 2013;6:579–603. doi: 10.3390/ph6050579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Lam JS, Huang H, Levitz SM. Effect of differential N-linked and O-linked mannosylation on recognition of fungal antigens by dendritic cells. PLoS One. 2007;2:e1009. doi: 10.1371/journal.pone.0001009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Lee K, Jin X, Zhang K, et al. A biochemical and pharmacological comparison of enzyme replacement therapies for the glycolipid storage disorder Fabry disease. Glycobiology. 2003;13:305–13. doi: 10.1093/glycob/cwg034. [DOI] [PubMed] [Google Scholar]
  105. Liu F, Wu X, Li L, et al. Use of baculovirus expression system for generation of virus-like particles: successes and challenges. Protein Expr Purif. 2013;90:104–16. doi: 10.1016/j.pep.2013.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Llop E, Gutierrez-Gallego R, Segura J, et al. Structural analysis of the glycosylation of gene-activated erythropoietin (epoetin delta, Dynepo) Anal Biochem. 2008;383:243–54. doi: 10.1016/j.ab.2008.08.027. [DOI] [PubMed] [Google Scholar]
  107. Loignon M, Perret S, Kelly J, et al. Stable high volumetric production of glycosylated human recombinant IFNalpha2b in HEK293 cells. BMC Biotechnol. 2008;8:65. doi: 10.1186/1472-6750-8-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. LUMIZYME® . (Alglucosidase alfa), for injection, for intravenous use [package insert] Cambridge, MA: Genzyme Corporation; 2010. [Google Scholar]
  109. LUVERIS® . (75 IU powder and solvent for solution for injection) [summary of product characteristics] London, UK: Merck Serono Europe Limited; 2005. [Google Scholar]
  110. MABTHERA® . (100 mg concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2008. [Google Scholar]
  111. Mahlangu J, Powell JS, Ragni MV, et al. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A. Blood. 2014;123:317–25. doi: 10.1182/blood-2013-10-529974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. McCue J, Kshirsagar R, Selvitelli K, et al. Manufacturing process used to produce long-acting recombinant factor VIII Fc fusion protein. Biologicals. 2015;43:213–9. doi: 10.1016/j.biologicals.2015.05.012. [DOI] [PubMed] [Google Scholar]
  113. McCue J, Osborne D, Dumont J, et al. Validation of the manufacturing process used to produce long-lasting recombinant factor IX Fc fusion protein. Haemophilia. 2014;20:e327–35. doi: 10.1111/hae.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Mei B, Chen Y, Chen J, et al. Expression of human coagulation factor VIII in a human hybrid cell line, HKB11. Mol Biotechnol. 2006;34:165–78. doi: 10.1385/MB:34:2:165. [DOI] [PubMed] [Google Scholar]
  115. MENVEO® . (powder and solution for solution for injection; meningococcal group A, C, W135 and Y conjugate vaccine) [summary of product characteristics] Siena, Italy: Novartis Vaccines and Diagnostics S.r.l; 2010. [Google Scholar]
  116. METALYSE® . (6,000 units; powder and solvent for solution for injection) [summary of product characteristics] Ingelheim am Rhein, Germany: Boehringer Ingelheim International GmbH; 2006. [Google Scholar]
  117. Mochizuki S, Hamato N, Hirose M, et al. Expression and characterization of recombinant human antithrombin III in Pichia pastoris . Protein Expr Purif. 2001;23:55–65. doi: 10.1006/prep.2001.1479. [DOI] [PubMed] [Google Scholar]
  118. MYALEPT™ . ([Metreleptin] for injection for subcutaneous use) [package insert] Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2014. [Google Scholar]
  119. MYOZYME® . (50 mg powder for concentrate for solution for infusion) [summary of product characteristics] Naarden, The Netherlands: Genzyme Europe B.V; 2011. [Google Scholar]
  120. Nagarajan T, Marissen WE, Rupprecht CE. Monoclonal antibodies for the prevention of rabies: theory and clinical practice. Antibody Technol J. 2014;4:1–12. [Google Scholar]
  121. NAGLAZYME® . (Galsulfase) injection for intravenous use [package insert] Novato, CA: BioMarin Pharmaceutical Inc; 2005. [Google Scholar]
  122. Noguchi A, Mukuria CJ, Suzuki E, et al. Immunogenicity of N-glycolylneuraminic acid-containing carbohydrate chains of recombinant human erythropoietin expressed in Chinese hamster ovary cells. J Biochem. 1995;117:59–62. doi: 10.1093/oxfordjournals.jbchem.a124721. [DOI] [PubMed] [Google Scholar]
  123. NOVOSEVEN® . (1.2 mg [60 KIU] powder and solvent for solution for injection) [summary of product characteristics] Bagsværd, Denmark: Novo Nordisk A/S; 2006. [Google Scholar]
  124. NOVOTHIRTEEN® . (2,500 IU powder and solvent for solution for injection) [summary of product characteristics] Bagsværd, Denmark: Novo Nordisk A/S; 2012. [Google Scholar]
  125. NPLATE® . (250 micrograms powder for solution for injection) [summary of product characteristics] Thousand Oaks, CA: Amgen Inc; 2009. [Google Scholar]
  126. NULOJIX® . (250 mg powder for concentrate for solution for infusion) [summary of product characteristics] Uxbridge, UK: Bristol-Myers Squibb Pharma EEIG; 2011. [Google Scholar]
  127. OBIZUR™ . ([Antihemophilic factor (recombinant), porcine sequence] lyophilized powder for solution for intravenous injection) [package insert] Westlake Village, CA: Baxter Healthcare Corporation; 2014. [Google Scholar]
  128. Octapharma. (2014). European Commission publishes approval of Octapharma’s human cell line recombinant FVIII (NUWIQ®) across all age groups in haemophilia A. Octapharma press release [online]. Available from: http://www.octapharma.com/en/about/newsroom/press-releases/news-single-view.html?tx_ttnews[tt_news]=528&cHash=041c2ff8ba682b310190e283d18d195d [last accessed 14 May 2014]
  129. Office of Device Evaluation, Center for Devices and Radiological Health. (2001). OP-1 Putty HDE Approval Letter Silver Spring, MD: US Food and Drug Administration [Google Scholar]
  130. OPGENRA® . (3.3 mg powder for implantation suspension) [summary of product characteristics] Dublin, Ireland: Olympus Biotech International Limited; 2014. [Google Scholar]
  131. ORENCIA® . (250 mg powder for concentrate for solution for infusion) [summary of product characteristics] Uxbridge, UK: Bristol-Myers Squibb Pharma EEIG; 2012. [Google Scholar]
  132. OVIDREL® . Pre-filled syringe (choriogonadotropic alfa injection) for subcutaneous use [package insert] Rockland, MA: EMD Serono, Inc; 2014. [Google Scholar]
  133. OVITRELLE® . (250 micrograms/0.5 mL solution for injection in pre-filled syringe) [summary of product characteristics] London, UK: Merck Serono Europe Limited; 2006. [Google Scholar]
  134. Padler-Karavani V, Yu H, Cao H, et al. Diversity in specificity, abundance, and composition of anti-Neu5Gc antibodies in normal humans: potential implications for disease. Glycobiology. 2008;18:818–30. doi: 10.1093/glycob/cwn072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Pauwels K, Herman P, Van Vaerenbergh B, et al. Animal cell cultures: risk assessment and biosafety recommendations. Appl Biosaf. 2007;12:26–38. [Google Scholar]
  136. PERJETA® . (420 mg concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2013a. [Google Scholar]
  137. PERJETA® . ([Pertuzumab] injection, for intravenous use) [package insert] South San Francisco, CA: Genentech, Inc; 2013b. [Google Scholar]
  138. Perot E, Enjolras N, Le Quellec S, et al. Expression and characterization of a novel human recombinant factor IX molecule with enhanced in vitro and in vivo clotting activity. Thromb Res. 2015;135:1017–24. doi: 10.1016/j.thromres.2015.02.034. [DOI] [PubMed] [Google Scholar]
  139. Peters RT, Low SC, Kamphaus GD, et al. Prolonged activity of factor IX as a monomeric Fc fusion protein. Blood. 2010;115:2057–64. doi: 10.1182/blood-2009-08-239665. [DOI] [PubMed] [Google Scholar]
  140. Peters RT, Toby G, Lu Q, et al. Biochemical and functional characterization of a recombinant monomeric factor VIII-Fc fusion protein. J Thromb Haemost. 2013;11:132–41. doi: 10.1111/jth.12076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Petricciani J, Sheets R. An overview of animal cell substrates for biological products. Biologicals. 2008;36:359–62. doi: 10.1016/j.biologicals.2008.06.004. [DOI] [PubMed] [Google Scholar]
  142. Picanco-Castro V, Biaggio RT, Cova DT, et al. Production of recombinant therapeutic proteins in human cells: current achievements and future perspectives. Protein Pept Lett. 2013;20:1373–81. doi: 10.2174/092986652012131112130322. [DOI] [PubMed] [Google Scholar]
  143. Powell JS, Josephson NC, Quon D, et al. Safety and prolonged activity of recombinant factor VIII Fc fusion protein in hemophilia A patients. Blood. 2012;119:3031–7. doi: 10.1182/blood-2011-09-382846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Powell JS, Pasi KJ, Ragni MV, et al. Phase 3 study of recombinant factor IX Fc fusion protein in hemophilia B. N Engl J Med. 2013;369:2313–23. doi: 10.1056/NEJMoa1305074. [DOI] [PubMed] [Google Scholar]
  145. PREVNAR® . (13 suspension for injection; pneumococcal polysaccharide conjugate vaccine [13-valent, adsorbed]) [summary of product characteristics] Sandwich, Kent: Pfizer Limited; 2009. [Google Scholar]
  146. PROCRIT® . (epoetin alfa) injection, for intravenous or subcutaneous use [package insert] Horsham, PA: Janssen Products, LP; 2000. [Google Scholar]
  147. PROLIA® . (60 mg solution for injection in a pre-filled syringe) [summary of product characteristics] Breda, The Netherlands: Amgen Europe B.V; 2010. [Google Scholar]
  148. PULMOZYME® . (Dornase alfa) inhalation solution [package insert] South San Francisco, CA: Genentech, Inc; 2010. [Google Scholar]
  149. Ranieri VM, Thompson BT, Barie PS, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366:2055–64. doi: 10.1056/NEJMoa1202290. [DOI] [PubMed] [Google Scholar]
  150. RAXIBACUMAB™ . (Injection, for intravenous use) [package insert] Reseach Triangle Park, NC: GlaxoSmithKline; 2014. [Google Scholar]
  151. REBIF® . (22 micrograms solution for injection in pre-filled syringe) [summary of product characteristics] London, UK: Merck Serono Europe Limited; 2008. [Google Scholar]
  152. REFACTO AF® . (250 IU powder and solvent for solution for injection) [summary of product characteristics] Sandwich, Kent: Pfizer Limited; 2014. [Google Scholar]
  153. REMICADE® . (100 mg powder for concentrate for solution for infusion) [summary of product characteristics] Leiden, The Netherlands: Janssen Biologics B.V; 2009. [Google Scholar]
  154. REOPRO® . Abciximab for intravenous administration [package insert] Indianapolis, IN: Eli Lilly and Company; 2013. [Google Scholar]
  155. REPLAGAL® . (1 mg/mL concentrate for solution for infusion) [summary of product characteristics] Danderyd, Sweden: Shire Human Genetic Therapies AB; 2006. [Google Scholar]
  156. REPLAGAL® . (Agalsidase alfa ghu) [package insert] North Ryde, Australia: Shire Australia Pty. Limited; 2010. [Google Scholar]
  157. RITUXAN® . (rituximab) injection, for intravenous use [package insert] South San Francisco, CA: Genentech, Inc; 2014. [Google Scholar]
  158. ROACTEMRA® . (20 mg/mL concentrate for solution for infusion) [summary of product characteristics] Shire Park, Welwyn Garden City: Roche Registration Limited; 2013. [Google Scholar]
  159. Rossi N, Silva BG, Astray R, et al. Effect of hypothermic temperatures on production of rabies virus glycoprotein by recombinant Drosophila melanogaster S2 cells cultured in suspension. J Biotechnol. 2012;161:328–35. doi: 10.1016/j.jbiotec.2012.05.016. [DOI] [PubMed] [Google Scholar]
  160. SAIZEN® . [Somatropin (rDNA origin) for injection] for subcutaneous injection [package insert] Rockland, MA: EMD Serono Inc; 1987. [Google Scholar]
  161. Scherer WF, Syverton JT, Gey GO. Studies on the propagation in vitro of poliomyelitis viruses. IV. Viral multiplication in a stable strain of human malignant epithelial cells (strain HeLa) derived from an epidermoid carcinoma of the cervix. J Exp Med. 1953;97:695–710. doi: 10.1084/jem.97.5.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Schiedner G, Hertel S, Bialek C, et al. Efficient and reproducible generation of high-expressing, stable human cell lines without need for antibiotic selection. BMC Biotechnol. 2008;8:13. doi: 10.1186/1472-6750-8-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Schlaeger EJ, Christensen K. Transient gene expression in mammalian cells grown in serum-free suspension culture. Cytotechnology. 1999;30:71–83. doi: 10.1023/A:1008000327766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. SEROSTIM® . [Somatropin (rDNA origin) for injection] for subcutaneous use [package insert] Rockland, MA: EMD Serono Inc; 1987. [Google Scholar]
  165. Shahrokh Z, Royle L, Saldova R, et al. Erythropoietin produced in a human cell line (Dynepo) has significant differences in glycosylation compared with erythropoietins produced in CHO cell lines. Mol Pharm. 2011;8:286–96. doi: 10.1021/mp100353a. [DOI] [PubMed] [Google Scholar]
  166. Shapiro AD, Ragni MV, Valentino LA, et al. Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients. Blood. 2012;119:666–72. doi: 10.1182/blood-2011-07-367003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. SIMPONI® . (50 mg solution for injection in pre-filled pen) [summary of product characteristics] Leiden, The Netherlands: Janssen Biologics B.V; 2009. [Google Scholar]
  168. SIMULECT® . (20 mg powder and solvent for solution for injection or infusion) [summary of product characteristics] Horsham, West Sussex: Novartis Europharm Limited; 2008. [Google Scholar]
  169. Somatropin Biopartners . (2 mg powder and solvent for prolonged-release suspension for injection) [summary of product characteristics] Reutlingen, Germany: BioPartners GmbH; 2013. [Google Scholar]
  170. Staudacher E, Altmann F, Wilson IB, et al. Fucose in N-glycans: from plant to man. Biochim Biophys Acta. 1999;1473:216–36. doi: 10.1016/s0304-4165(99)00181-6. [DOI] [PubMed] [Google Scholar]
  171. STELARA® . (45 mg solution for injection) [summary of product characteristics] Beerse, Belgium: Janssen-Cilag International NV; 2013. [Google Scholar]
  172. Swiech K, de Freitas MC, Covas DT, et al. Recombinant glycoprotein production in human cell lines. Methods Mol Biol. 2015;1258:223–40. doi: 10.1007/978-1-4939-2205-5_12. [DOI] [PubMed] [Google Scholar]
  173. Swiech K, Kamen A, Ansorge S, et al. Transient transfection of serum-free suspension HEK 293 cell culture for efficient production of human rFVIII. BMC Biotechnol. 2011;11:114. doi: 10.1186/1472-6750-11-114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Swiech K, Picanco-Castro V, Covas DT. Human cells: new platform for recombinant therapeutic protein production. Protein Expr Purif. 2012;84:147–53. doi: 10.1016/j.pep.2012.04.023. [DOI] [PubMed] [Google Scholar]
  175. SYLVANT®. (2015). ([Siltuximab] for injection, for intravenous infusion) [package insert]. Horsham, PA; Janssen Biotech Inc [Google Scholar]
  176. SYLVANT™ . (100 mg powder for concentrate for solution for infusion) [summary of product characteristics] Beerse, Belgium: Janssen-Cilag International NV; 2014. [Google Scholar]
  177. SYNAGIS® . (50 mg powder and solvent for solution for injection) [summary of product characteristics] Maidenhead, UK: AbbVie Ltd; 2009. [Google Scholar]
  178. TANZEUM™ . ([Albiglutide] for injection, for subcutaneous use) [package insert] Wilmington, DE: GlaxoSmithKline, LLC; 2014. [Google Scholar]
  179. tbo-filgrastim . (Injection for subcutaneous use) [package insert] Vilnius, Lithuania: Sicor Biotech UAB; 2012. [Google Scholar]
  180. Tekoah Y, Tzaban S, Kizhner T, et al. Glycosylation and functionality of recombinant beta-glucocerebrosidase from various production systems. Biosci Rep. 2013;33:e00071. doi: 10.1042/BSR20130081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Thomas P, Smart TG. HEK293 cell line: a vehicle for the expression of recombinant proteins. J Pharmacol Toxicol Methods. 2005;51:187–200. doi: 10.1016/j.vascn.2004.08.014. [DOI] [PubMed] [Google Scholar]
  182. THYROGEN® . (0.9 mg powder for solution for injection) [summary of product characteristics] Naarden, The Netherlands: Genzyme Europe B.V; 2010. [Google Scholar]
  183. TNKASE® . (Tenecteplase) [package insert] South San Francisco, CA: Genentech, Inc; 2011. [Google Scholar]
  184. TRETTEN® . Coagulation factor XIII A-subunit (recombinant) for intravenous use. Lyophilized powder for solution for injection [prescribing information] Bagsvaerd, Denmark: Novo Nordisk A/S; 2014. [Google Scholar]
  185. TRULICITY™ . ([Dulaglutide] injection, for subcutaneous use) [package insert] Indianapolis, IN: Eli Lilly and Company; 2014. [Google Scholar]
  186. TYSABRI® . (300 mg concentrate for solution for infusion) [summary of product characteristics] Research Triangle Park, NC: Biogen Idec Inc; 2011. [Google Scholar]
  187. US Food and Drug Administration. (2010). CDER new molecular entity (NME) & new BLA calendar year approvals. Available from: http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologicApprovalReports/UCM260585.pdf [last accessed 31 Dec 2010]
  188. US Food and Drug Administration. (2011). CDER new molecular entity (NME) & new BLA calendar year approvals. Available from: http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologicApprovalReports/UCM276989.pdf [last accessed 31 Dec 2011]
  189. US Food and Drug Administration. (2012). CDER new molecular entity (NME) & new BLA calendar year approvals. Available from: http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologicApprovalReports/UCM342733.pdf [last accessed 31 Dec 2012]
  190. US Food and Drug Administration. (2013). CDER new molecular entity (NME) & new BLA calendar year approvals. Available from: http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologicApprovalReports/NDAandBLAApprovalReports/UCM406851.pdf [last accessed 31 Dec 2013]
  191. US Food and Drug Administration. (2014). US Food and Drug Administration. Drugs@FDA. Original new drug application (NDA and BLA) approvals. February–May. Available from: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm. [last accessed on 8 Dec 2014]
  192. Vectibix® . (20 mg/mL concentrate for solution for infusion) [summary of product characteristics] Breda, The Netherlands: Amgen Europe B.V; 2014. [Google Scholar]
  193. Vedder AC, Linthorst GE, Houge G, et al. Treatment of Fabry disease: outcome of a comparative trial with agalsidase alfa or beta at a dose of 0.2 mg/kg. PLoS One. 2007;2:e598. doi: 10.1371/journal.pone.0000598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Victoza® . (6 mg/mL solution for injection in pre-filled pen) [summary of product characteristics] Bagsværd, Denmark: Novo Nordisk A/S; 2009. [Google Scholar]
  195. VIMIZIM® . ([Elosulfase alfa] injection, for intravenous use) [package insert] Novato, CA: BioMarin Pharmaceutical Inc; 2014a. [Google Scholar]
  196. VIMIZIM® . (1 mg/mL concentrate for solution for infusion) [summary of product characteristics] London, UK: BioMarin Europe Limited; 2014b. [Google Scholar]
  197. Vink T, Oudshoorn-Dickmann M, Roza M, et al. A simple, robust and highly efficient transient expression system for producing antibodies. Methods. 2014;65:5–10. doi: 10.1016/j.ymeth.2013.07.018. [DOI] [PubMed] [Google Scholar]
  198. VORAXAZE® . ([Glucarpidase] for injection, for intravenous use) [package insert] West Conshohocken, PA: BTG International Inc; 2012. [Google Scholar]
  199. VPRIV® . (400 units powder for solution for infusion) [summary of product characteristics] Dublin, Ireland: Shire Pharmaceuticals Ireland Limited; 2010a. [Google Scholar]
  200. VPRIV® . (Velaglucerase alfa for injection) [package insert] Cambridge, MA: Shire Human Genetic Therapies, Inc; 2010b. [Google Scholar]
  201. VPRIV® . ([Velaglucerase alfa for injection] for intravenous use) [package insert] Lexington, MA: Shire Human Genetic Therapies, Inc; 2013. [Google Scholar]
  202. World Health Organization. (2013). Guidelines on the quality, safety, and efficacy of biotherapeutic protein products prepared by recombinant DNA technology: replacement of annex 3 of WHO technical report series, No. 814 [Google Scholar]
  203. Wraith JE. Enzyme replacement therapy with idursulfase in patients with mucopolysaccharidosis type II. Acta Paediatr Suppl. 2008;97:76–8. doi: 10.1111/j.1651-2227.2008.00661.x. [DOI] [PubMed] [Google Scholar]
  204. Wurm FM. Production of recombinant protein therapeutics in cultivated mammalian cells. Nat Biotechnol. 2004;22:1393–8. doi: 10.1038/nbt1026. [DOI] [PubMed] [Google Scholar]
  205. Wysham C, Blevins T, Arakaki R, et al. Efficacy and safety of dulaglutide added onto pioglitazone and metformin versus exenatide in type 2 diabetes in a randomized controlled trial (AWARD-1) Diabetes Care. 2014;37:2159–67. doi: 10.2337/dc13-2760. [DOI] [PubMed] [Google Scholar]
  206. XEOMIN® . (Incobotulinumtoxin A) for injection, for intramuscular use [package insert] Greensboro, NC: Merz Pharmaceuticals, LLC; 2014. [Google Scholar]
  207. XIAFLEX® . (Collagenase clostridium histolyticum) for injection, for intralesional use [package insert] Chesterbrook, PA: Auxilium Pharmaceuticals, Inc; 2014. [Google Scholar]
  208. XIGRIS® . ([Drotrecogin alfa (activated)] injection, powder, lyophilized, for solution for intravenous use) [package insert] Indianapolis, IN: Eli Lilly and Company; 2008. [Google Scholar]
  209. XOLAIR® . (75 mg powder and solvent for solution for injection) [summary of product characteristics] Horsham, West Sussex: Novartis Europharm Limited; 2010. [Google Scholar]
  210. Xu X, Nagarajan H, Lewis NE, et al. The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat Biotechnol. 2011;29:735–41. doi: 10.1038/nbt.1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. XYNTHA® . [Antihemophilic factor (recombinant), plasma/albumin-free] for intravenous use freeze-dried powder [package insert] Philadelphia, PA: Wyeth Pharmaceuticals Inc; 2011. [Google Scholar]
  212. Yamaguchi K, Itoh K, Ohnishi N, et al. Engineered long terminal repeats of retroviral vectors enhance transgene expression in hepatocytes in vitro and in vivo. Mol Ther. 2003;8:796–803. doi: 10.1016/j.ymthe.2003.08.005. [DOI] [PubMed] [Google Scholar]
  213. Yang WC, Lu J, Kwiatkowski C, et al. Perfusion seed cultures improve biopharmaceutical fed-batch production capacity and product quality. Biotechnol Prog. 2014a;30:616–25. doi: 10.1002/btpr.1884. [DOI] [PubMed] [Google Scholar]
  214. Yang WC, Lu J, Nguyen NB, et al. Addition of valproic acid to CHO cell fed-batch cultures improves monoclonal antibody titers. Mol Biotechnol. 2014b;56:421–8. doi: 10.1007/s12033-013-9725-x. [DOI] [PubMed] [Google Scholar]
  215. YERVOY® . (5 mg/mL concentrate for solution for infusion) [summary of product characteristics] Uxbridge, UK: Bristol-Myers Squibb Pharma EEIG; 2011. [Google Scholar]
  216. ZALTRAP® . (25 mg/mL concentrate for solution for infusion) [summary of product characteristics] Paris, France: sanofi-aventis groupe; 2013a. [Google Scholar]
  217. ZALTRAP® . ([ziv-Aflibercept] injection for intravenous infusion) [package insert] Bridgewater, NJ: sanofi-aventis U.S. LLC; 2013b. [Google Scholar]
  218. ZEVALIN® . (1.6 mg/mL kit for radiopharmaceutical preparations for infusion) [summary of product characteristics] Amsterdam, The Netherlands: Spectrum Pharmaceuticals B.V; 2009. [Google Scholar]
  219. Zimran A, Pastores GM, Tylki-Szymanska A, et al. Safety and efficacy of velaglucerase alfa in Gaucher disease type 1 patients previously treated with imiglucerase. Am J Hematol. 2013;88:172–8. doi: 10.1002/ajh.23383. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Critical Reviews in Biotechnology are provided here courtesy of Taylor & Francis

RESOURCES